US20170192000A1 - Method for monitoring cathepsin s inhibition - Google Patents

Method for monitoring cathepsin s inhibition Download PDF

Info

Publication number
US20170192000A1
US20170192000A1 US15/456,353 US201715456353A US2017192000A1 US 20170192000 A1 US20170192000 A1 US 20170192000A1 US 201715456353 A US201715456353 A US 201715456353A US 2017192000 A1 US2017192000 A1 US 2017192000A1
Authority
US
United States
Prior art keywords
cathepsin
white blood
antibody
blood cells
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/456,353
Inventor
Ana Patricia BENTO PEREIRA DA SILVA
Sebastian Dziadek
Barbara Ecabert
Michael Gerg
Everson Nogoceke
Moritz Marcinowski
Bernhard Reis
Thomas Schindler
Michel Achille Edouard THERON
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Publication of US20170192000A1 publication Critical patent/US20170192000A1/en
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AGE
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROCHE DIAGNOSTICS GMBH
Assigned to ROCHE DIAGNOSTICS GMBH reassignment ROCHE DIAGNOSTICS GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DZIADEK, SEBASTIAN, GERG, MICHAEL, MARCINOWSKI, MORITZ
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOGOCEKE, EVERSON, SCHINDLER, THOMAS, REIS, BERNHARD, ECABERT, BARBARA, BENTO PEREIRA DA SILVA, Ana Patricia, THERON, MICHEL ACHILLE EDOURAD
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70539MHC-molecules, e.g. HLA-molecules
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/81Protease inhibitors
    • G01N2333/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • G01N2333/8139Cysteine protease (E.C. 3.4.22) inhibitors, e.g. cystatin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96466Cysteine endopeptidases (3.4.22)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Definitions

  • the tissue sample is blood.
  • APCs Antigen Presenting cells
  • MHC antigenic peptides as (MHC) class II-antigenic peptide complex
  • APCs comprise dendritic cells, monocytes, macrophages and B cells.
  • MHC II polypeptide refers to the alpha chain and the beta chain constituting the MHC II complex.
  • An example for a method to measure a polypeptide is an ELISA. This type of protein quantitation is based on an antibody capable of capturing a specific antigen, and a second antibody capable of detecting the captured antigen. Methods for preparation and use of antibodies, and the assays mentioned hereinbefore are described in Harlow, E. and Lane, D. Antibodies: A Laboratory Manual, (1988), Cold Spring Harbor Laboratory Press.
  • WO 2010/121918 discloses Cathepsin S inhibitors which can be tested in the method of the present invention.
  • FIG. 2 Detection of Ii p10 expression on B cells using a neoepitope antibody.
  • PBMCs isolated from blood donation of a healthy donor (R172) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1 or vehicle.
  • Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using two different neoepitope antibodies, namely 7B8 (black) and 13C4 (grey).
  • Stain index Median Fluorescence Intensity (MFI) B cells /MFI Tcells
  • FIG. 4 Cathepsin S inhibitor IC50 concentrations.
  • PBMCs isolated from blood donations of eleven healthy donors (R39; R185; R198; R204; R209; R140; R154; R175; R213; R4;R214) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1, Cathepsin S inhibitor 2 or vehicle.
  • Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using 7B8 neoepitope antibody.
  • Response curves and IC50 values have been calculated with GraphPad Prism software.
  • FIG. 8A-8D Cohort B data, see FIG. 7 description.
  • 1 ⁇ Cell Satining Buffer (BioLegend, order ID: 420201. Storage: +4 C the 1 ⁇ Cell Satining Buffer is ready to use.
  • 1 ⁇ PBS without CaCl 2 and without MgCl 2 (Gibco, order ID: 14190-094). Storage: room temperature. The PBS is ready to use. 5 ml Polystyrene round-bottom tube (FACS tube, BD, order ID: 352052). Storage: room temperature.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Wood Science & Technology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention relates to a method for monitoring Cathepsin S inhibitor activity in tissue samples of animals comprising:
    • a) providing a tissue sample of an animal to whom a Cathepsin S inhibitor has been administered or providing a tissue sample of an animal that was contacted in vitro with a Cathepsin S inhibitor, wherein the tissue samples comprise white blood cells,
    • b) measuring li p10 peptide level in white blood cells of the tissue sample of step a) by flow cytometry and
    • c) correlating li p10 peptide level in white blood cells to Cathepsin S inhibitor dose, wherein a Cathepsin S inhibitor leads to increased level of li p10 peptide in the white blood cells.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to PCT Application No. PCT/EP2015/071002, filed on Sep. 15, 2015, which claims priority to European patent application no. 14185409.1, filed on Sep. 18, 2004, the entire contents of which is incorporated herein by reference.
  • SUBMISSION OF SEQUENCE LISTING ON ASCII TEXT FILE
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Feb. 23, 2017 is named P32272-US_SeqListing.txt and is 9,793 bytes in size.
  • BACKGROUND OF THE INVENTION
  • The present invention provides an antibody directed to li p10 peptide and its use in methods for monitoring Cathepsin S inhibition.
  • Cathepsin S is well known for its critical function in the proteolytic digestion of the invariant chain chaperone molecules, thus controlling antigen presentation to CD4+ T-cells by major histocompatibility complex (MHC) class II molecules or to NK1.1+ T-cells via CD1 molecules. Cathepsin S also appears to participate in direct processing of exogenous antigens for presentation by MHC class II to CD4+ T-cells, or in cross-presentation by MHC class I molecules to CD8+ T-cells. In addition, in its secreted form Cathepsin S is implicated in degradation of the extracellular matrix, which may contribute to the pathology of a number of diseases, including arthritis, atherosclerosis and chronic obstructive pulmonary disease. Therefore, inhibition of Cathepsin S is a promising target for the development of novel therapeutics for a variety of indications.
  • MHC class II molecules bind and display antigenic peptides as class II-peptide complexes on the cell surface of antigen presenting cells (APCs) for recognition by CD4+ helper cells. The molecular mechanisms leading to formation of class II-peptide complexes and the presentation of antigens on the cell surface begin with synthesis of class II αβ heterodimers in the endoplasmatic reticulum. During biosynthesis these αβ heterodimers associate with a type II membrane protein, the isoforms of the invariant chain li, to form a nonameric complex of αβli. The αβli complex traverses the Golgi apparatus and is delivered to intracellular compartments. In these acidic compartments, the li is degraded and finally liberated from the αβli complexes, allowing class II molecules to bind antigenic peptides. The proteolytical processing of li results in shorter fragments. The cysteine protease Cathepsin S plays a key role in processing li in APCs. Cathepsin S degrades the invariant chain li from class II-li complexes generating class II-CLIP. Hence, inhibition of Cathepsin S leads to a decrease of CLIP peptide and to accumulation of the precursor fragment, i.e. the invariant chain li degradation peptide p10 (li p10).
  • EP 1 315 966 discloses a method for monitoring the effect of in vivo administration of cathepsin S inhibitors. The method includes the purification of white blood cells from a blood sample of a subject treated with a Cathepsin S inhibitor, preparation of whole cell lysates of the purified white blood cells and analyzing the whole cell lysates for presence of li p10 fragment of invariant chain li. This method requires blood samples of about 10-30 ml and is labour intensive (isolation of white blood cells and preparation of whole cell lysates). The detection of li p10 is done by ELISA or Western blot.
  • The aim of the present invention is to provide an assay to monitor Cathepsin S inhibition which overcomes at least some of the drawbacks of the prior art methods.
  • In a first object, the present invention provides a method for monitoring Cathepsin S inhibitor activity in tissue samples of animals comprising:
  • a) providing a tissue sample of an animal to whom a Cathepsin S inhibitor has been administered or providing a tissue sample of an animal that was contacted in vitro with a Cathepsin S inhibitor, wherein the tissue sample comprises white blood cells,
  • b) measuring lip10 peptide level in white blood cells of the tissue sample of step a) by flow cytometry and
  • c) correlating li p10 peptide level in white blood cells to Cathepsin S inhibitor dose, wherein a Cathepsin S inhibitor leads to increased level of li p10 peptide in white blood cells.
  • In a second object the present invention provides a method for the identification of a Cathepsin S inhibitor comprising:
  • a) treating a non-human animal with a test compound,
  • b) providing a tissue sample of the animal of step a) comprising white blood cells,
  • c) measuring lip10 peptide level in the white blood cells of the tissue sample of step b),
  • wherein an increased level of li p10 peptide in white blood cells in the sample of step a) compared to a level of li p10 peptide in white blood cells in a tissue sample of an untreated animal is indicative for a Cathepsin S inhibitor.
  • In a preferred embodiment of the methods of the present invention the tissue sample is blood.
  • In a further preferred embodiment of the methods of the present invention the white blood cells are Antigen Presenting Cells (APCs), preferably the APCs are selected from B cells, monocytes, macrophages and dendritic cells, more preferably selected from B cells and monocytes.
  • The term Antigen Presenting cells (APCs) as used herein refers to cells that display antigenic peptides as (MHC) class II-antigenic peptide complex on its surface. APCs comprise dendritic cells, monocytes, macrophages and B cells.
  • In a third object, the present invention provides an anti-li p10 antibody comprising:
  • a VH domain comprising a CDR1 comprising the amino acid sequence of Seq. Id. No. 4, a CDR2 comprising the amino acid sequence of Seq. Id. No. 5, CDR3 sequence comprising the amino acid sequence of Seq. Id. No. 6., and a VL domain comprising a CDR1 comprising the amino acid sequence of Seq. Id. No. 7, a CDR2 comprising the amino acid sequence of Seq. Id. No. 8, a CDR3 sequence comprising the amino acid sequence of Seq. Id. No. 9.
  • In a particular embodiment of the present invention, the VH domain of the anti-li p10 antibody comprises the amino acid sequence of Seq. Id. No. 10 and the VL domain of the anti-li p10 antibody comprises the amino acid sequence of Seq. Id. No. 11.
  • The inventive anti-li p10 specific antibody is referred to herein as 7B8 neoepitope antibody originating from Oryctologous cuniculus. This antibody is characterized by CDRs and VH, VL sequences disclosed in Seq. Id. Nos. 4-11.
  • The term Cathepsin S refers to a cysteine protease in the papain super-family. The amino acid sequence of human Cathepsin S is given in Seq. Id. No. 1.
  • The term “invariant chain li” refers to invariant polypeptide of major histocompatibility complex or CD74. The amino acid sequence of human invariant chain li is given in Seq. Id. No. 2.
  • The term li p10 peptide refers to class II-associated li peptide. The amino acid sequence of human li p10 is given in Seq. Id. No. 3.
  • The term “MHC II polypeptide” as used herein refers to the alpha chain and the beta chain constituting the MHC II complex.
  • The term animal as used herein comprises human beings and non-human animals such as e.g. monkey, dog, pig, rabbit, guinea pig and rodents.
  • The term “compound” is used herein in the context of a “test compound” or a “drug candidate compound” described in connection with the assays of the present invention. As such, these compounds comprise organic or inorganic compounds, derived synthetically or from natural sources. The compounds include inorganic or organic compounds such as polynucleotides, lipids or hormone analogs that are characterized by relatively low molecular weights. Other biopolymeric organic test compounds include peptides comprising from about 2 to about 40 amino acids and larger polypeptides comprising from about 40 to about 500 amino acids, such as antibodies or antibody conjugates.
  • The inventors of the present invention have surprisingly found that li p10 peptide can be detected in white blood cells. The methods of the present invention have the advantage compared to the methods described in the prior art that tissue samples, preferably whole blood, can be assayed directly without involving the isolation of white blood cells. In the methods of the present invention there is no need for the isolation of e.g. white blood cells from whole blood samples, in order to be able to be processed in a method of the present invention. This is of great importance if the assay is used in clinical settings where a large number of samples have to be processed. The methods of the present invention require smaller tissue samples, preferably blood samples, than the prior art methods.
  • Methods for detection and/or measurement of polypeptides in biological samples are well known in the art and include, but are not limited to, Western-blotting, Flow cytometry, ELISAs or RIAs, or various proteomics techniques. Monoclonal or polyclonal antibodies recognizing the li p10 peptide or fragments thereof, can either be generated for the purpose of detecting the polypeptides or peptide fragments, e.g. by immunizing rabbits with purified proteins, or known antibodies recognizing the polypeptides or peptide fragments can be used. For example, an antibody capable of binding to the denatured proteins, such as a polyclonal antibody, can be used to detect LI P10 peptide or MHC II polypeptide in a Western Blot. An example for a method to measure a polypeptide is an ELISA. This type of protein quantitation is based on an antibody capable of capturing a specific antigen, and a second antibody capable of detecting the captured antigen. Methods for preparation and use of antibodies, and the assays mentioned hereinbefore are described in Harlow, E. and Lane, D. Antibodies: A Laboratory Manual, (1988), Cold Spring Harbor Laboratory Press.
  • A preferred method for the detection of li p10 peptide is flow cytometry. Flow cytometry methods are described in Handbook of Flow Cytometry Method, J. Paul Robinson (Editor); Flow Cytometry—A Basic Introduction, Michael G Ormerod (2008) and Current Protocols in Cytometry (2010), Wiley. In this method proteins are not denatured but preserved in their native form.
  • WO 2010/121918 discloses Cathepsin S inhibitors which can be tested in the method of the present invention.
  • SHORT DESCRIPTION OF THE FIGURES
  • FIG. 1: Detection of Ii p10 expression on B cells and T cells using a neoepitope antibody. PBMCs isolated from blood donation of a healthy donor (R213) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1 or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using a different neoepitope antibody 7B8 (black).
  • FIG. 2: Detection of Ii p10 expression on B cells using a neoepitope antibody. PBMCs isolated from blood donation of a healthy donor (R172) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1 or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using two different neoepitope antibodies, namely 7B8 (black) and 13C4 (grey). Stain index=Median Fluorescence Intensity (MFI)B cells/MFITcells
  • FIG. 3 A: Ii p10 accumulates in B cells upon treatment with Cathepsin S inhibitors. PBMCs isolated from blood donations of eleven healthy donors (R39; R185; R198; R204; R209; R140; R154; R175; R213; R4;R214) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1 or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using 7B8 neoepitope antibody. Stain index=MFIB cells/MFITcells.
  • FIG. 3 B: Ii p10 accumulates in B cells upon treatment with Cathepsin S inhibitors. PBMCs isolated from blood donations of eleven healthy donors (R39; R185; R198; R204; R209; R140; R154; R175; R213; R4;R214) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 2 or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using 7B8 neoepitope antibody. Stain index=MHB cells/MFITcells.
  • FIG. 4: Cathepsin S inhibitor IC50 concentrations. PBMCs isolated from blood donations of eleven healthy donors (R39; R185; R198; R204; R209; R140; R154; R175; R213; R4;R214) were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1, Cathepsin S inhibitor 2 or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using 7B8 neoepitope antibody. Response curves and IC50 values have been calculated with GraphPad Prism software.
  • FIGS. 5 A-F: Ii p10 neoepitope assay longitudinal variability. PBMCs isolated from blood donations of eleven healthy donors (R198; R209; R154; R213; R214) at two time points and were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1, or vehicle. Ii p10 detection on B cells (CD20+) and T cells (CD3+) was determined by flow cytometry using 7B8 neoepitope antibody.
  • FIG. 6 A: Ii p10 accumulation in B cells is detected by the neoepitope assay. Ii p10 detection on B cells (CD20+) was determined by flow cytometry using 7B8 neoepitope antibody clone. PBMCs isolated from 3 cynos (Macaca fascicularis) blood donations were incubated for 20 h with increasing concentrations of Cathepsin S inhibitor 1 or vehicle. The p10 stain index of Macaca fascicularis samples was determined by dividing the Median Fluorescence Intensity (MFI) of B cells by the MFI of Forward Scatter Low CD20 negative (FSCloCD20−) cells.
  • FIG. 6 B: Ii p10 accumulation in B cells is detected by the neoepitope assay after a single oral dose of Cathepsin S inhibitor. Ii p10 detection on B cells (CD20+) was determined by flow cytometry using 7B8 neoepitope antibody clone. Blood was collected from animals 2 h and 7 h after a single dose of 50 mg/kg. Ii p10 neoepitope assay was performed in whole blood. The p10 stain index of Macaca fascicularis samples was determined by dividing the Median Fluorescence Intensity (MFI) of B cells by the MFI of Forward Scatter Low CD20 negative (FSCloCD20−) cells.
  • FIG. 7A-7D: Quantification of p10 on samples obtained from subjects participating in a single center, randomized, double-blind, placebo-controlled, single ascending dose study to evaluate the safety, tolerability, pharmacokinetic and pharmacodynamic effects of single doses of Cathepsin inhibitor 1 in healthy male and female volunteers. To account for and limit the impact of the expected inter-subject variability as observed in preclinical experiments, an interleaved cohort (“leapfrog”) design was employed. Subjects were recruited into two cohorts of eight subjects each (Cohorts A and B). For each individual within a cohort, the study was a randomized, placebo controlled, four treatment, four periods, four-way crossover. The single dose applied shortly after time point 0 hours is indicated on each FIG. 7A to 7D (cohort A) and FIG. 8A to 8D (cohort B).
  • FIG. 8A-8D: Cohort B data, see FIG. 7 description.
  • EXPERIMENTAL PART
  • In Vitro Assay with Cathepsin S Inhibitor
      • Use a sterile Polystyrene 24 well plate
      • Add 500 μl complete RPMI+Cathepsin S inhibitor with increasing concentrations (0 to 10 μM)
      • Add 500 μl of PBMC suspension (2×106) to each well
      • Mix the content of the wells and incubate the plate for 20 h at +37° C., 5% CO2 in a humidified incubator
    FACS Staining:
      • prepare 1× lyse/fix solution
      • add 4 mL of pre-warmed 1× lyse/fix solution per 2×106 PBMCs or 200 μl blood
      • incubate for 10 min at 37 C, wash the cells 1× with PBS (250×g, 5 minutes, RT), remove supernatant
      • add 2 mL of 1× permeabilising solution per tube
      • incubate for 20 min at RT
      • wash the cells 2× with 2 ml Cell Stain Buffer (250×g, 5 minutes, RT), remove supernatant
      • add 10 μg/mL unlabeled anti-p10 antibody in Cell Staining Buffer for 1 h at RT
      • wash the cells 1× with 2 ml Cell Stain Buffer (250×g, 5 minutes, RT), remove supernatant
      • add 1:125 PE-conjugated goat anti-rabbit IgG antibody in Cell Staining for 30 min at RT in the dark
      • wash the cells 2× with 2 ml Cell Stain Buffer (250×g, 5 minutes, RT), remove supernatant
      • add the antibodies (comp. control) or antibody mixture (diluted in 100 μl/sample Cell Stain Buffer) to each tube
      • incubate for 20 min at +4° C. in the dark
      • wash the cells 1× with 2 ml Cell Stain Buffer (250×g, 5 minutes, RT), remove supernatant
      • resuspend pellet in 200 μl Cell Stain Buffer and keep the cells in the dark until the analysis on the FACS
  • Antibodies:
  • goat anti-rabbit IgG PE SouthernBiotech Cat# 4050-09
    1:125/sample
    CD3 Brilliant Violet 421 BioLegend Cat# 300434
    5 μl/sample
    CD14 APC Beckman Coulter Cat# IM2580
    5 μl/sample
    CD20 AlexaFluor 488 BD Biosciences Cat# 558056
    20 μl/sample
    CD45 PerCP/Cy5.5 BioLegend Cat# 304028
    2 μl/sample
  • Materials and Buffers:
  • 24-well plates, BD, order ID: 351147Storage: room temperature, sterile, polystyrene non tissue culture treated
  • 10× BD Phosphoflow Lyse/Fix buffer lysis buffer (BD, order ID: 558049). Storage: +4° C. Dilute the 5× BD Phosphoflow Lyse/Fix buffer with H2O bidest. to 1× and prewarm it to the required temperature prior to use 10× Phosflow Perm Buffer IV permeabilisation buffer (BD, order ID: 560746). Storage at RT. Dilute 10× BD Phosphoflow Perm Buffer IV with PBS to 1×.
  • 1× Cell Satining Buffer (BioLegend, order ID: 420201. Storage: +4 C the 1× Cell Satining Buffer is ready to use. 1× PBS without CaCl2 and without MgCl2 (Gibco, order ID: 14190-094). Storage: room temperature. The PBS is ready to use. 5 ml Polystyrene round-bottom tube (FACS tube, BD, order ID: 352052). Storage: room temperature.
  • Amino Acid Sequences
  • Sequence name Seq. Id. No.
    Human Cathepsin S 1
    Human invariant chain li 2
    Human li p10 3
    VH CDR 1 of anti li p10 antibody 7B8 4
    VH CDR 2 of anti li p10 antibody 7B8 5
    VH CDR 3 of anti li p10 antibody 7B8 6
    VL CDR 1 of anti li p10 antibody 7B8 7
    VL CDR 2 of anti li p10 antibody 7B8 8
    VL CDR 3 of anti li p10 antibody 7B8 9
    VH chain of anti li p10 antibody 7B8 10
    VL chain of anti li p10 antibody 7B8 11

Claims (20)

1. A method for monitoring Cathepsin S inhibitor activity in a tissue sample of animals comprising:
a) providing a tissue sample of an animal to whom a Cathepsin S inhibitor has been administered or providing a tissue sample of an animal that was contacted in vitro with a Cathepsin S inhibitor, wherein the tissue sample comprise white blood cells;
b) measuring lip10 peptide level in white blood cells of the tissue sample of step a) by flow cytometry; and
c) correlating li p10 peptide level in white blood cells to Cathepsin S inhibitor dose, wherein a Cathepsin S inhibitor leads to increased level of li p10 peptide in white blood cells.
2. A method for the identification of a Cathepsin S inhibitor comprising:
a) contacting a non-human animal with a test compound;
b) providing a tissue sample of the animal of step a) comprising white blood cells; and
c) measuring li p10 peptide level in white blood cells of the tissue sample of step b) by flow cytometry;
wherein an increased level of li p10 peptide in white blood cells in the sample of step a) compared to a level of li p10 peptide in white blood cells in a tissue sample of an untreated animal is indicative for a Cathepsin S inhibitor.
3. The method of claim 1 wherein the tissue sample is blood.
4. The method of claim 2 wherein the tissue sample is blood.
5. The method of claim 1, wherein the white blood cells are Antigen Presenting Cells (APCs).
6. The method of claim 2, wherein the white blood cells are Antigen Presenting Cells (APCs).
7. The method of claim 5, wherein the APCs are selected from B cells and monocytes.
8. The method of claim 6, wherein the APCs are selected from B cells and monocytes.
9. The method of claim 1 wherein the animal is a human being.
10. The method of claim 2, wherein the non-human animal is a rodent.
11. The method of claim 1, wherein the white blood cells are permeabilized to measure li p10 peptide by flow cytometry.
12. The method of claim 2, wherein the white blood cells are permeabilized to measure li p10 peptide by flow cytometry.
13. The method of claim 1, wherein an antibody directed to li p10 is used in flow cytometry to measure li p10 in white blood cells.
14. The method of claim 2, wherein an antibody directed to li p10 is used in flow cytometry to measure li p10 in white blood cells.
15. An anti li p10 antibody comprising a VH domain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:4, a CDR2 comprising the amino acid sequence of SEQ ID NO:5, CDR3 sequence comprising the amino acid sequence of SEQ ID NO:6., and a VL domain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:7, a CDR2 comprising the amino acid sequence of SEQ ID NO:8, a CDR3 sequence comprising the amino acid sequence of SEQ ID NO:9.
16. The anti li p10 antibody of claim 9, wherein the VH domain comprises the amino acid sequence of SEQ ID NO:10 and the VL domain comprises the amino acid sequence of SEQ ID NO:11.
17. The method of claim 13, wherein said antibody is the antibody of claim 15.
18. The method of claim 13, wherein said antibody is the antibody of claim 16.
19. The method of claim 14, wherein said antibody is the antibody of claim 15.
20. The method of claim 14, wherein said antibody is the antibody of claim 16.
US15/456,353 2014-09-18 2017-03-10 Method for monitoring cathepsin s inhibition Abandoned US20170192000A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP14185409.1 2014-09-18
EP14185409 2014-09-18
PCT/EP2015/071002 WO2016041915A1 (en) 2014-09-18 2015-09-15 Method for monitoring cathepsin s inhibition

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/071002 Continuation WO2016041915A1 (en) 2014-09-18 2015-09-15 Method for monitoring cathepsin s inhibition

Publications (1)

Publication Number Publication Date
US20170192000A1 true US20170192000A1 (en) 2017-07-06

Family

ID=51584978

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/456,353 Abandoned US20170192000A1 (en) 2014-09-18 2017-03-10 Method for monitoring cathepsin s inhibition

Country Status (5)

Country Link
US (1) US20170192000A1 (en)
EP (1) EP3194610B1 (en)
JP (1) JP6826527B2 (en)
CN (1) CN106715472A (en)
WO (1) WO2016041915A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7026132B2 (en) * 2000-09-06 2006-04-11 Ortho-Mcneil Pharmaceutical, Inc. Method of monitoring the effect of Cathepsin S inhibitors
WO2012041824A1 (en) * 2010-10-01 2012-04-05 F. Hoffmann-La Roche Ag Method for monitoring cathepsin s inhibition

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1212839C (en) * 2000-09-06 2005-08-03 奥索-麦克尼尔药品公司 Method for treating allergies using substituted pyrazoles
EP1315741A2 (en) * 2000-09-06 2003-06-04 Ortho-McNeil Pharmaceutical, Inc. A method for treating allergies
US7326719B2 (en) * 2003-03-13 2008-02-05 Boehringer Ingelheim Pharmaceuticals, Inc. Cathepsin S inhibitors
JP5518997B2 (en) 2009-04-20 2014-06-11 エフ.ホフマン−ラ ロシュ アーゲー Proline derivatives as cathepsin inhibitors
EP2670440B1 (en) * 2011-02-01 2018-09-05 Genmab A/S Human antibodies and antibody-drug conjugates against cd74

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7026132B2 (en) * 2000-09-06 2006-04-11 Ortho-Mcneil Pharmaceutical, Inc. Method of monitoring the effect of Cathepsin S inhibitors
WO2012041824A1 (en) * 2010-10-01 2012-04-05 F. Hoffmann-La Roche Ag Method for monitoring cathepsin s inhibition

Also Published As

Publication number Publication date
WO2016041915A1 (en) 2016-03-24
EP3194610A1 (en) 2017-07-26
EP3194610B1 (en) 2019-11-13
JP2017529531A (en) 2017-10-05
JP6826527B2 (en) 2021-02-03
CN106715472A (en) 2017-05-24

Similar Documents

Publication Publication Date Title
Meng et al. Definition of the nature and hapten threshold of the β-lactam antigen required for T cell activation in vitro and in patients
US20230041030A1 (en) Antigen-binding proteins targeting shared neoantigens
US7094555B2 (en) Methods of MHC class II epitope mapping, detection of autoimmune T cells and antigens, and autoimmune treatment
US20160024176A1 (en) Ox40l fusion proteins and uses thereof
EP3045468B1 (en) Novel peptides and their use in diagnosis and treatment
US20220373538A1 (en) Composition for determination of cell-mediated immune responsiveness
Barberá et al. APL-1, an altered peptide ligand derived from human heat-shock protein 60, selectively induces apoptosis in activated CD4+ CD25+ T cells from peripheral blood of rheumatoid arthritis patients
IL175063A (en) Use of pif peptides in the manufacture of a medicament for treating intolerance to embryo implantation
Uhl et al. In situ cell surface proteomics reveals differentially expressed membrane proteins in retinal pigment epithelial cells during autoimmune uveitis
Seedhom et al. Protein translation activity: a new measure of host immune cell activation
US9994817B2 (en) Use of ligands for the programmed cell death receptor conjugated to solid supports for cultivating human regulatory T cells
US8841421B2 (en) S100A9 interaction screening method
Haskins et al. Dendritic cell-mediated responses to secreted Cryptosporidium effectors promote parasite-specific CD8+ T cell responses
US20170192000A1 (en) Method for monitoring cathepsin s inhibition
US8222211B2 (en) Methods of administering PIF agonist peptides and uses thereof
Haskins et al. Dendritic cell-mediated responses to secreted Cryptosporidium effectors are required for parasite-specific CD8+ T cell responses
Holm et al. Confirmation of a disease model of pemphigus vulgaris: characterization and correlation between disease parameters in 90 mice
EP2436776A1 (en) Method for monitoring Cathepsin S inhibition
US12006373B1 (en) Identification and evaluation of novel peptide ligands specific to human CD3 epsilon
EP4246143A1 (en) In-vitro method for diagnosis of pemphigus vulgaris and/or of determining disease activity in a pemphigus vulgaris patient
TWI845803B (en) Anti-ccr8 antibodies and uses thereof
Zhang et al. Vam6 upregulated by lactic acid inhibits anti-tumor effects of intratumoral iNKT cells via modulating AMPK/mTOR pathways
Hasan et al. SARS-CoV-2 infection induces adaptive NK cell responses by spike protein-mediated induction of HLA-E expression
Morel Mechanism underpinning the immunosuppressive effects of the mycobacterial macrolide mycolactone
CA3241439A1 (en) Off-target prediction method for antigen-recognition molecules binding to mhc-peptide targets

Legal Events

Date Code Title Description
AS Assignment

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BENTO PEREIRA DA SILVA, ANA PATRICIA;ECABERT, BARBARA;NOGOCEKE, EVERSON;AND OTHERS;SIGNING DATES FROM 20141007 TO 20141017;REEL/FRAME:044932/0957

Owner name: ROCHE DIAGNOSTICS GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DZIADEK, SEBASTIAN;GERG, MICHAEL;MARCINOWSKI, MORITZ;REEL/FRAME:044933/0049

Effective date: 20141110

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AGE;REEL/FRAME:044933/0174

Effective date: 20141201

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ROCHE DIAGNOSTICS GMBH;REEL/FRAME:044933/0087

Effective date: 20141201

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION